Next Article in Journal
Thermodynamic Biomarkers of Neuroinflammation: Nanothermometry, Energy–Stress Dynamics, and Predictive Entropy in Glial–Vascular Networks
Previous Article in Journal
Spatial- and Phospho-Proteomic Profiling Reveals Pancreatic and Hepatic Dysfunction in a Rat Model of Lethal Insulin Overdose
Previous Article in Special Issue
Defactinib in Combination with Mitotane Can Be an Effective Treatment in Human Adrenocortical Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Coenzyme Q10 and Intracellular Signalling Pathways: Clinical Relevance

Pharma Nord (UK) Ltd., Morpeth NE61 2DB, Northumberland, UK
Int. J. Mol. Sci. 2025, 26(22), 11024; https://doi.org/10.3390/ijms262211024
Submission received: 28 September 2025 / Revised: 11 November 2025 / Accepted: 11 November 2025 / Published: 14 November 2025
(This article belongs to the Special Issue Signalling Pathways in Metabolic Diseases and Cancers)

Abstract

Intracellular signalling pathways provide a mechanism to connect events at a cell surface to the nucleus and are of fundamental importance to normal cell functioning. Intracellular signalling pathways control many aspects of cell metabolism, including mitochondrial function, oxidative stress, inflammation, and apoptosis/ferroptosis. Randomised controlled clinical trials supplementing coenzyme Q10 (CoQ10) have reported significant clinical improvements in a number of disorders, in turn associated with the action of CoQ10 to promote normal mitochondrial function, reduce oxidative stress and inflammation, and mediate apoptosis and ferroptosis. However, the precise mechanisms by which CoQ10 facilitates beneficial changes in the above factors is not completely understood. In the present article, the evidence we have reviewed provides a supporting rationale that the beneficial role of CoQ10 in the above disorders occurs via mediation of major intracellular signalling pathways, including the Nrf2/NQO1, NF-κB, P13/AKT/mTOR, MAPK, JAK/STAT, WNT/B-catenin, AMPK-YAP-OPA1, and hedgehog (Hh) pathways; the clinical consequences of such mediation are also reviewed.

1. Introduction

Intracellular signalling pathways, also known as signal transduction cascades, essentially connect events at the cell surface to the nucleus and are of fundamental importance to normal cell functioning. In a typical pathway, binding of an extracellular molecule to specific receptors on the cell membrane results in the activation of enzymes such as kinases or phosphatases, in turn generating transcription factors that bind to DNA in the nucleus; in this way gene expression is regulated in response to external stimuli [1]. More than 300 individual pathways have been identified in human cells, although these can be broadly classified into less than a dozen general types, depending on the type of cell membrane receptor and mechanism of signal transduction [2]. Examples of external stimuli activating intracellular signalling pathways include hormones and growth factors, such as the binding of epidermal growth factor to its cell surface receptor, triggering the P13/AKT pathway to promote cell proliferation and survival, as detailed in Section 4 of this article.
Coenzyme Q10 (CoQ10) is usually described as a vitamin-like substance, although by definition, CoQ10 is not a vitamin since it is synthesised by most tissues within the human body. CoQ10 has a number of vital functions within cells, particularly in cellular energy production within mitochondria, but also as an antioxidant [3]. Deficiency of CoQ10 has been implicated in the pathogenesis of a number of disorders, and randomised controlled clinical trials supplementing CoQ10 in such disorders have demonstrated significant clinical improvements [4]. These symptomatic improvements are in turn linked with the action of CoQ10 to promote normal mitochondrial function, reduce oxidative stress and inflammation, and mediate apoptosis and ferroptosis [4]. However the precise mechanisms by which CoQ10 facilitates beneficial changes in the above factors are not completely understood. To further elucidate these mechanisms, in the present article we have therefore reviewed evidence for the interaction of CoQ10 with intracellular signalling pathways, since these are known to control many of the factors outlined above, and dysregulation of these pathways has been linked with the pathogenesis of a wide range of disorders [5]. Thus in this article, we have reviewed the potential benefits (for example, in reducing oxidative stress or inflammation) and clinical consequences of supplementary CoQ10 in mediating some of the major intracellular signalling pathways, including the Nrf2/NQO1, NF-κB, P13/AKT/mTOR, MAPK, JAK/STAT, WNT/B-catenin, AMPK-YAP-OPA1, and hedgehog (Hh) pathways.

2. The Nrf2/NQO1 Pathway

The Nrf2/NQO1 pathway is a vital defence mechanism, protecting cells from the damaging effects of oxidative stress induced by free radicals, as well as from other types of potentially damaging molecular species. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor which acts as a master regulator for cytoprotective gene expression, controlling the production of antioxidant enzymes, detoxification proteins, and other molecules responsible for protecting cells from the effects of harmful substances [6]. The action of Nrf2 is tightly regulated to prevent excessive or insufficient responses to cell stress. Nrf2 is kept inactive in the cytoplasm by a protein called Keap1. When cells encounter oxidative stress (or some other types of stress), Nrf2 is released from Keap1 and translocates to the nucleus, where it binds to specific DNA sequences called antioxidant response elements (AREs), activating the transcription of genes encoding antioxidant and detoxification proteins [7]. In addition, Nrf2 has a role in reducing inflammation by modulating the production of inflammatory molecules and promoting the resolution of inflammatory responses [8]. Nrf2 also plays a role in mitochondrial function, including mitochondrial biogenesis, mitophagy, and autophagy, all of which are essential for maintaining healthy mitochondria [9]. Nrf2 plays a role in stem cell biology, influencing the balance between stem cell self-renewal and differentiation [10].
NAD(P)H quinone dehydrogenase 1 (NQO1) is a Phase II detoxification enzyme that catalyses the two-electron reduction in quinones (and other compounds) to hydroquinones, preventing them from participating in harmful redox cycling, thereby preventing the formation of reactive oxygen free radical species (ROS). NQO1 detoxifies various quinones, including those found in the environment and those produced within the body such as vitamin K. A common polymorphism in the NQO1 gene (C609T) results in a non-functional protein. Individuals homozygous for this polymorphism lack NQO1 activity. NQO1 polymorphisms can affect an individual’s susceptibility to certain diseases and their response to environmental toxins [11].
There are several potential downstream targets of activated Nrf2; in addition to NQO1, these include the enzyme HO1 (haem oxygenase), the products of which have antioxidant, anti-inflammatory, and anti-apoptotic action. Reduced HO-1 levels have been associated with a number of disorders, including miscarriage and pre-eclampsia. Other downstream targets of activated Nrf2 include the antioxidant enzymes glutathione peroxidase, superoxide dismutase, and thioredoxin reductase [12].
The ability of CoQ10 to activate the Nrf2/NQO1 pathway by upregulating the expression of NQO1 and other antioxidant enzymes has implications for various health conditions. For example in diabetes, CoQ10’s ability to activate this pathway and reduce oxidative stress may be beneficial in managing diabetic complications [13]. Additionally, NQO1’s role in stabilizing p53, a tumour suppressor protein, suggests a broader role for CoQ10 in cancer prevention and treatment [14]. In the context of spinal cord injury, CoQ10 has been shown to suppress oxidative stress and apoptosis by activating this pathway, leading to increased levels of Nrf2 and NQO1 and reduced levels of p65, a component of the NF-κB pathway [15]. The above studies represent only a few examples of the action of CoQ10 in activating the Nrf2 pathway; a more complete list of such studies is summarised in Table 1. In every study, CoQ10 administration resulted in increased Nrf2 levels, usually accompanied by increased levels of antioxidant enzymes/reduced levels of oxidative stress, and reduced levels of inflammation and apoptosis.

3. The NF-κB Pathway

The nuclear factor kappa-light-chain-enhancer of the activated B cells (NF-κB) pathway is vital for regulating the immune response and inflammation. The NF-κB pathway is activated by diverse stimuli (including stress signals and pathogens) and plays a vital role in both the innate and adaptive immune responses. NF-κB comprises a family of transcription factors that regulate gene expression. The pathway involves the activation of NF-κB dimers, which then translocate to the nucleus to regulate the expression of genes involved in the immune response, inflammation, and apoptosis. NF-κB also has a role in cell proliferation, cell cycle regulation, and neuronal function [38].
CoQ10 can modulate the NF-κB pathway, resulting in reduced inflammation, oxidative stress, and apoptosis. Examples of suppression of the NF-κB pathway by CoQ10 include the reduction in inflammation induced by beta-amyloid in nerve cells [39], and the reduction in oxidative stress and apoptosis following spinal cord injury [15]. A more comprehensive summary of the effects of CoQ10 on the NF-κB pathway is given in Table 2; in every study listed, NF-κB was downregulated, typically accompanied by reductions in oxidative stress, inflammation, or apoptosis. It is of note that NF-κB can regulate the biosynthesis of CoQ10 by binding to specific sites in the COQ7 gene, indicating a feedback loop between CoQ10 and NF-κB [40].

4. The PI3K/AKT/mTOR Pathway

The P13K/AKT/mTOR pathway is an intracellular signalling pathway involved in various cell processes, including the promotion of cell survival by inhibiting apoptosis, the stimulation of cell growth and proliferation, and the regulation of glucose metabolism. PI3K refers to phosphoinositide 3-kinases, a family of lipid kinases activated by growth factors and other extracellular signals, which phosphorylate phosphatidylinositol lipids to generate second messengers such as PIP3. AKT refers to protein kinase B, a serine/threonine kinase that plays a central role in downstream signalling that is activated by PIP3 (phosphatidylinositol 3,4,5-trisphosphate). mTOR (mammalian target of rapamycin) is a protein kinase that acts as a downstream effector of AKT, integrating signals from various pathways; it essentially acts as a master regulator of protein synthesis and cell growth [61].
CoQ10 can both activate or inhibit the P13K/AKT/mTOR pathway, depending on the specific cellular context, including cell type or type of cellular stress. For example, in some instances, CoQ10 can promote autophagy by inactivating the PI3K/AKT/mTOR pathway, and in other situations CoQ10 can inhibit autophagy by activating the PI3K/AKT/mTOR pathway [62]; in the latter study, the activation of pancreatic stellate cells was inhibited by suppressing autophagy, leading to anti-fibrotic effects. In other examples, activation of the pathway by CoQ10 has been shown to promote wound healing [63], or to protect against beta-amyloid-induced neuronal death [64]. Inhibition of the pathway by CoQ10 can protect cells from damage caused by heat stress [65] and inhibit the formation of osteoclast cells responsible for bone resorption, thereby preventing osteoporosis [66]. The PI3K/Akt pathway is also involved in stem cell aging, and CoQ10 can inhibit this pathway in mesenchymal stem cells, potentially mitigating cellular aging [67].

5. The MAPK Pathway

The MAPK pathway (mitogen-activated protein kinase pathway) is involved in cell growth, differentiation, proliferation, and in stress response. MAPK comprises several related pathways, including the ERK/MAPK pathway activated by growth factors, and the JNK/p38 MAPK pathway activated by stress signals. As with other pathways, binding of extracellular molecular stimuli at specific cell surface receptors results in the activation of the mitogen-activated protein kinase enzymes, which translocate to the nucleus to activate specific genes. Aberrant activation of MAPK pathways is frequently observed in various cancers, and the MAPK pathway is also implicated in inflammatory and neuropathic pain [68].
As with the P13K/AKT/mTOR above, CoQ10 can both activate and inhibit the MAPK pathway. For example, CoQ10 can inhibit the MAPK pathway to reduce inflammation; in rat articular chondrocytes, CoQ10 was found to prevent the interleukin-1 beta-induced inflammatory response by inhibiting the MAPK signalling pathway [69]. CoQ10 also prevented RANKL-induced osteoclastogenesis by promoting autophagy via inactivation of the PI3K/AKT/mTOR and MAPK pathways [70]. In a rat model of spermatogenic dysfunction induced by a high-fat diet, supplementary CoQ10 suppressed testicular dysfunction via inhibition of the MAPK pathway [71]. Alternatively CoQ10 can activate the MAPK pathway to improve memory and neuronal differentiation; in a study investigating the effects of CoQ10 on Alzheimer’s disease in rats, CoQ10 was found to improve memory and neuronal differentiation, partly by activating the TLR-4/MAPK pathway [72]. In chicken myocardial cells, CoQ10 upregulated HSP heat shock protein via the MAPK pathway, demonstrating its role in cellular stress response [73].

6. The JAK/STAT Pathway

The JAK-STAT pathway plays a role in the immune response by regulating cytokine signalling, which is essential for inflammation and immune cell development; the pathway is also involved in cell growth, differentiation, and survival. The key components of the pathway are Janus kinases and signal transducers and activators of transcription (STATs). Binding of extracellular molecules (cytokines or growth factors) to specific receptors at the cell surface results in activation of Janus kinases; STAT proteins are in turn activated, translocating to the nucleus, binding to specific DNA regions to activate gene transcription. The pathway is tightly regulated, with feedback mechanisms, such as the suppressor of cytokine signalling proteins, which help to terminate the signal. Dysregulation of the JAK-STAT pathway is implicated in various diseases, including cancers (e.g., leukaemia, lymphoma), autoimmune disorders (e.g., rheumatoid arthritis, inflammatory bowel disease), and other inflammatory conditions [74].
There is evidence that CoQ10 can mediate the JAK/STAT pathway, particularly its role in inflammatory responses. An in silico study identified CoQ10-inducible genes that are functionally connected to the JAK/STAT signalling pathway [75].

7. The Wnt/β-Catenin Pathway

The Wnt/β-catenin pathway is a cell-to-cell signalling pathway involved in various biological processes, including embryonic development, tissue homeostasis, cell proliferation, differentiation, and cell polarity. The pathway can be activated through canonical (β-catenin-dependent) or non-canonical (β-catenin-independent) pathways, depending on the cellular context. Wnt refers to extracellular proteins that bind to specific cell surface receptors, in turn activating β-catenin, an intracellular signalling protein that translocates to the nucleus, activating the transcription of target genes involved in cell proliferation, survival, differentiation, and migration.
Dysregulation of the Wnt/β-catenin pathway, often through mutations or epigenetic changes, can lead to uncontrolled cell proliferation and is implicated in various cancers. Aberrant activation of this pathway is also associated with developmental disorders, neurodegenerative diseases, and bone diseases [76].
Research indicates that CoQ10 can play a protective role in neurodegenerative diseases by modulating the Wnt/β-catenin pathway. In a preclinical study examining the effects of cyclophosphamide-induced chemobrain, CoQ10 was found to reduce neuronal apoptosis and preserve hippocampal neurogenesis by influencing the Wnt/β-catenin signalling pathway [77]. In addition, in a preclinical rat model of renal necroinflammation, supplementary CoQ10 attenuated renal fibrosis via mediation of the Wnt/β-catenin pathway [78]. The Wnt/β-catenin pathway plays a fundamental role in cardiac regeneration and tissue repair; this pathway has a key role in promoting cardiomyocyte proliferation during neonatal cardiac development and is re-activated in response to cardiac injury [79]. In this regard the role of supplemental CoQ10 in promoting normal cardiovascular function is of note [80], although direct interaction between CoQ10 and the Wnt/β-catenin signalling pathway in the context of cardiac regeneration has not been reported to date.

8. The AMPK-YAP-OPA1 Pathway

The AMPK-YAP-OPA1 pathway is a signalling pathway involved in regulating mitochondrial function, energy metabolism, and cell survival; in effect it provides a mechanism for cells to adapt to changing energy demands. AMPK is an AMP-activated protein kinase, a cellular energy sensor that regulates metabolism that is activated under conditions of cellular stress such as low energy or nutrient deprivation. YAP, or Yes-associated protein, is a transcriptional co-activator involved in cell growth and proliferation. OPA1, or optic atrophy 1, is a protein responsible for mitochondrial fusion and maintaining mitochondrial integrity [81].
A preclinical study in mice provided evidence that CoQ10 can activate the AMPK-YAP-OPA1 pathway to improve mitochondrial function, reduce oxidative stress, and improve energy metabolism, thereby reducing atherosclerosis [82].

9. The Hedgehog Pathway

The hedgehog (Hh) pathway is a cell-to-cell communication pathway that plays a key role in organogenesis, stem cell maintenance, and tissue repair, both in embryonic development and in adult tissue homeostasis. So-called hedgehog ligands bind to specific cell surface receptors (PTCH); this in turn activates a protein (SMO), which then activates transcription factors (GLI) in the nucleus. Dysregulation of the Hh pathway is implicated in several diseases, notably cancer, where it can contribute to tumour initiation, progression, and resistance to therapy. Hh inhibitors (HHIs), such as vismodegib and sonidegib, are targeted therapies that block the Hh pathway, helping to slow or stop cancer cell growth [83].
In patients with advanced basal cell carcinoma treated with Hh pathway inhibitors, CoQ10 supplementation can reduce the need for dose reductions due to the side effects of medications such as vismodegib and sonidegib, particularly muscle spasms and fatigue [84].

10. Discussion

One might first question why there are so many individual intracellular signalling pathways performing broadly similar functions; however, there is an argument that having multiple pathways that can achieve similar outcomes provides a level of redundancy. If one pathway is compromised, others can compensate, ensuring the cell can still carry out essential functions. In addition, each intracellular pathway does not operate in isolation; the pathways interact through mechanisms such as cross-talk [85] or scaffolding proteins [86]. Such interactions between the various pathways allow cells to fine-tune an appropriate response to a wide range of external stimuli. An example is the interaction between the P13/AKT/mTOR and MAPK pathways, both of which have key roles in the regulation of cell growth, proliferation, and survival. There are several points at which these pathways interact, facilitating coordinated control of downstream targets; thus, following activation the MAPK pathway can phosphorylate and activate components of the PI3K/AKT/mTOR pathway, amplifying the signalling output [87].
The intracellular signalling pathways described in this article have a number of features in common. Firstly, they provide a mechanism for altered gene expression in response to extracellular stimuli. Secondly, these pathways are involved (although by differing mechanisms) in cell growth, differentiation and survival, immune response, and inflammation. Thirdly, dysregulation of these pathways has been implicated in the pathogenesis of a range of disorders; particularly in cancer, but also in disorders as diverse as neurodegenerative disease and osteoporosis. Fourthly, there is evidence that CoQ10 can promote the normal functioning of all of these pathways. In particular, there is considerable evidence from preclinical studies for the role of CoQ10 in the activation of the Nrf2 pathway and inhibition of the NF-κB pathway, as summarised in Table 1 and Table 2, respectively. Dysregulation of intracellular signalling pathways can occur for a number of reasons, including mitochondrial dysfunction and oxidative stress. Given the role of CoQ10 in promoting normal mitochondrial function and as an antioxidant, it follows that CoQ10 has the potential to ameliorate the conditions leading to pathway dysfunction; in addition, there is evidence that CoQ10 can beneficially affect the action of various components in each of these pathways.
Dysregulation of signalling pathways can have the following clinical consequences: (i) Dysregulation of pathways involved in cell growth, proliferation, and apoptosis can result in uncontrolled cell growth and tumour formation. (ii) Signalling pathways are essential for immune cell activation, differentiation, and function; pathway dysregulation can result in immune deficiency and increased risk of infection, as well as autoimmune disease. (iii) Signalling pathways are involved in neurotransmitter release, receptor function, and neuronal communication; pathway dysfunction can result in neurological disorders. (iv) Signalling pathways regulate metabolism, including glucose uptake and lipid metabolism; pathway dysregulation can result in metabolic disorders like diabetes and obesity. (v) Dysfunction of signalling pathways involved in regulating blood pressure, heart contractility, and blood vessel function can contribute to the development of cardiovascular disease. The clinical consequences of intracellular signalling pathway dysfunction has been summarised in Table 3.
As noted above, many of the intracellular signalling pathways described in this article are involved in the development of cancer; in this regard the potential role of supplemental CoQ10 in the prevention or treatment of cancer has been reviewed by Mantle et al. [88]. It is also of note that CoQ10 interacts with peroxisome proliferator-activated receptor (PPAR)-related pathways, in the regulation of mitochondrial function, oxidative stress, inflammation, and lipid metabolism. CoQ10 first activates the AMPK pathway, resulting in increased expression of PPAR$\alpha$ and PPAR$\gamma$, leading to improved mitochondrial function, improved lipid metabolism, and reduced inflammation [89].
Table 3. Clinical consequences of intracellular signalling pathway dysfunction.
Table 3. Clinical consequences of intracellular signalling pathway dysfunction.
PathwayCellular Function of PathwayPrincipal Clinical Consequence of Pathway Dysfunction
Nrf2/NQO1/Protection against oxidative stressNeurodegenerative disorders, including Parkinson’s disease, Alzheimer’s disease, multiple sclerosis, and amyotrophic lateral sclerosis [90]
Nf-κBRegulation of immune response and inflammationAutoimmune disorders, including rheumatoid arthritis, lupus erythematous, and IBS [91]
P13K/AKT/mTORRegulation of cell growth and proliferationCancer, including breast, colon, and skin cancers [92]
MAPKControl of cell growth, differentiation and survivalCancer, including colon, pancreatic, lung, and skin cancers [93]
JAK/STATRegulation of immune response and inflammation; cell growth and proliferationAutoimmune disorders (e.g., rheumatoid arthritis, IBS) and cancer (e.g., haematological cancers, breast and lung cancer) [94]
Wnt/β-cateninNormal embryonic development; control of cell growthEmbryonic abnormalities; cancer [95]
AMPK/YAP/OPA1Control of energy metabolismCardiovascular disease; metabolic disorders (diabetes, obesity) [96]
HedgehogNormal embryonic developmentDevelopmental disorders (e.g., craniofacial defects); cancer (e.g., basal cell carcinoma) [97]
As noted in Table 3, dysregulation of the Nrf2 pathway is principally associated with neurodegenerative disorders; reduced activity of the Nrf2 pathway is associated with Alzheimer’s disease [98], Parkinson’s disease [99], amyotrophic lateral sclerosis [100], Huntington’s disease [101], and Friedreich’s ataxia [102]. However, it is of note that in addition to the principal disorders listed in Table 3, most of the pathways have been implicated in a variety of other disorders. As an example, in addition to neurodegenerative disorders associated with reduced activity of the Nrf2/NQO1 pathway, other disorders that have been linked with the reduced activity of this pathway include cardiovascular disease [103], hepatic disease [104], renal disease [105], pulmonary disease [106], gastrointestinal disease [107], and musculoskeletal disease [108].
Intracellular signalling pathways are therapeutic targets in a variety of disorders. Thus some types of cancer therapy may target intracellular pathways that drive uncontrolled cell growth: examples include inhibitors of the MAPK or P13/AKT/mTOR pathways, such as trametinib for melanoma and everolimus for breast cancer [93,109]. Further examples of medicines, either approved or undergoing clinical trials, targeting intracellular signalling pathways have been summarised in Table 4.
It is also of note that the relevance of intracellular signalling pathways in clinical chemistry has become increasingly recognised, with components of the various pathways acting as potential biomarkers for the diagnosis and monitoring of a number of diseases. Examples of intracellular signalling pathway components used as clinical biomarkers include RAS proteins for the early detection of cancer and SMAD proteins for the diagnosis of immune disorders such as multiple sclerosis and Crohn’s disease [131,132]. However, in general terms, the use of intracellular signalling pathway components as clinical biomarkers for the diagnosis or prognosis of disease remains an area still in the relatively early stages of research.
An area of relevance to intracellular signalling pathways is hormesis—a process in which a low dose of a stressor, which would be harmful at higher doses, triggers a beneficial response within the body. Hormesis promotes human health by activating the body’s defence and repair mechanisms, and hormesis activated by drugs or natural compounds is emerging as a promising preventive and therapeutic strategy in many chronic diseases [133,134]. An example of hormesis is the activation of the Nrf2 pathway, resulting in upregulation of cytoprotective antioxidant enzymes. In this regard, CoQ10 could be considered as a hormetic drug as it is able to induce health benefits at moderate doses (typically of the order of 100–200mg/day in clinical studies) by activating antioxidant pathways (Nrf2/NQO1) or inhibiting inflammatory pathways (NF-κB) in order to prevent or attenuate detrimental consequences of oxidative stress and chronic inflammation. However, it is of note that very high doses (up to 3000 mg/day) of CoQ10 used in some clinical studies reported no significant adverse effects [135].
In summary, in this article the importance of intracellular signalling pathways in the pathogenesis of a wide range of disorders has been highlighted. Supplementation with CoQ10 has been shown to be of benefit in a number of such disorders [88,136,137,138,139], although the mechanisms involved are not completely understood. In this article, the evidence we have reviewed provides a supporting rationale that the beneficial role of CoQ10 in these disorders occurs via mediation of intracellular signalling pathways, as summarised in Figure 1. Intracellular signalling pathways have primary effects in the form of first-line responses, such as the activation of protein kinases, and secondary effects such as the switching on or off of genes, or changes in cell metabolism. It is apparent from the data listed in Table 1 and Table 2 that supplementary CoQ10 has both primary and secondary effects on intracellular signalling pathways. A limitation of the present article is that most of the studies reviewed were preclinical; relatively few clinical studies have been carried out to specifically investigate the mediation of intracellular signalling pathways by CoQ10. As will be noted from Table 4, there is a precedent for developing novel medicines targeting intracellular signalling pathways, such as CoQ10, and further research into the clinical applications of the latter is now warranted.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analysed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

D.M. is a medical advisor at Pharma Nord (UK) Ltd.

References

  1. Antebi, Y.E.; Nandagopal, N.; Elowitz, M.B. An operational view of intercellular signaling pathways. Curr. Opin. Syst. Biol. 2017, 1, 16–24. [Google Scholar] [CrossRef]
  2. Valdespino-Gómez, V.M.; Valdespino-Castillo, P.M.; Valdespino-Castillo, V.E. Cell signaling pathways interaction in cellular proliferation: Potential target for therapeutic interventionism. Cirugía Cir. 2015, 83, 165–174. [Google Scholar] [CrossRef]
  3. Crane, F.L. Biochemical functions of coenzyme Q10. J. Am. Coll. Nutr. 2001, 20, 591–598. [Google Scholar] [CrossRef] [PubMed]
  4. Mantle, D.; Turton, N.; Hargreaves, I.P. Depletion and supplementation of Coenzyme Q10 in Secondary Deficiency Disorders. Front. Biosci. Landmark Ed. 2022, 27, 322. [Google Scholar] [CrossRef] [PubMed]
  5. Xue, C.; Chu, Q.; Shi, Q.; Zeng, Y.; Lu, J.; Li, L. Wnt signaling pathways in biology and disease: Mechanisms and therapeutic advances. Signal Transduct. Target. Ther. 2025, 10, 106. [Google Scholar] [CrossRef] [PubMed]
  6. Ngo, V.; Duennwald, M.L. Nrf2 and oxidative stress: A general overview of mechanisms and implications in human disease. Antioxidants 2022, 11, 2345. [Google Scholar] [CrossRef]
  7. Baird, L.; Yamamoto, M. The Molecular Mechanisms Regulating the KEAP1-NRF2 Pathway. Mol. Cell Biol. 2020, 40, e00099-20. [Google Scholar] [CrossRef]
  8. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules. 2020, 25, 5474. [Google Scholar] [CrossRef]
  9. Esteras, N.; Abramov, A.Y. Nrf2 as a regulator of mitochondrial function: Energy metabolism and beyond. Free Radic. Biol. Med. 2022, 189, 136–153. [Google Scholar] [CrossRef]
  10. Dodson, M.; Anandhan, A.; Zhang, D.D.; Madhavan, L. An NRF2 Perspective on stem cells and ageing. Front. Aging 2021, 2, 690686. [Google Scholar] [CrossRef]
  11. Zhou, Y.Y.; Sun, J.H.; Wang, L.; Cheng, Y.Y. Genetic polymorphism of NQO1 influences susceptibility to coronary heart disease in a chinese population: A cross-sectional study and meta-anaylsis. Pharmgenom. Pers. Med. 2023, 16, 825–833. [Google Scholar] [CrossRef]
  12. Loboda, A.; Damulewicz, M.; Pyza, E.; Jozkowicz, A.; Dulak, J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell. Mol. Life Sci. 2016, 73, 3221–3247. [Google Scholar] [CrossRef] [PubMed]
  13. Samimi, F.; Baazm, M.; Nadi, Z.; Dastghaib, S.; Rezaei, M.; Jalali-Mashayekhi, F. Evaluation of antioxidant effects of Coenzyme Q10 against hyperglycemia-mediated oxidative stress by focusing on Nrf2/Keap1/HO-1 signaling pathway in the liver of diabetic rats. Iran. J. Med. Sci. 2024, 49, 661–670. [Google Scholar] [CrossRef] [PubMed]
  14. Asher, G.; Lotem, J.; Kama, R.; Sachs, L.; Shaul, Y. NQO1 stabilizes p53 through a distinct pathway. Proc. Natl. Acad. Sci. USA 2002, 99, 3099–3104. [Google Scholar] [CrossRef] [PubMed]
  15. Li, X.; Zhan, J.; Hou, Y.; Chen, S.; Hou, Y.; Xiao, Z.; Luo, D.; Lin, D. Coenzyme Q10 suppresses oxidative stress and apoptosis via activating the Nrf-2/NQO-1 and NF-κB signaling pathway after spinal cord injury in rats. Am. J. Transl. Res. 2019, 11, 6544–6552. [Google Scholar]
  16. Choi, H.K.; Pokharel, Y.R.; Lim, S.C.; Han, H.K.; Ryu, C.S.; Kim, S.K.; Kwak, M.K.; Kang, K.W. Inhibition of liver fibrosis by solubilized coenzyme Q10: Role of Nrf2 activation in inhibiting transforming growth factor-beta1 expression. Toxicol. Appl. Pharmacol. 2009, 240, 377–384. [Google Scholar] [CrossRef]
  17. Pala, R.; Orhan, C.; Tuzcu, M.; Sahin, N.; Ali, S.; Cinar, V.; Atalay, M.; Sahin, K. Coenzyme Q10 supplementation modulates NFκB and Nrf2 pathways in exercise training. J. Sports Sci. Med. 2016, 15, 196–203. [Google Scholar]
  18. Yang, H.L.; Lin, M.W.; Korivi, M.; Wu, J.J.; Liao, C.H.; Chang, C.T.; Liao, J.W.; Hseu, Y.C. Coenzyme Q0 regulates NFκB/AP-1 activation and enhances Nrf2 stabilization in attenuation of LPS-induced inflammation and redox imbalance: Evidence from in vitro and in vivo studies. Biochim. Biophys. Acta. 2016, 1859, 246–261. [Google Scholar] [CrossRef]
  19. Li, L.; Du, J.; Lian, Y.; Zhang, Y.; Li, X.; Liu, Y.; Zou, L.; Wu, T. Protective effects of Coenzyme Q10 against hydrogen peroxide-Induced oxidative stress in PC12 cell: The role of Nrf2 and antioxidant enzymes. Cell. Mol. Neurobiol. 2016, 36, 103–111. [Google Scholar] [CrossRef]
  20. Kabel, A.M.; Elkhoely, A.A. Ameliorative effect of Coenzyme Q10 and/or Candesartan on carboplatin-Induced nephrotoxicity: Roles of apoptosis, transforming growth factor-Β1, Nuclear Factor Kappa-B and the Nrf2/HO-1 pathway. Asian Pac. J. Cancer Prev. 2017, 18, 1629–1636. [Google Scholar] [CrossRef]
  21. Khodir, A.E.; Atef, H.; Said, E.; ElKashef, H.A.; Salem, H.A. Implication of Nrf2/HO-1 pathway in the coloprotective effect of coenzyme Q10 against experimentally induced ulcerative colitis. Inflammopharmacology 2017, 25, 119–135. [Google Scholar] [CrossRef] [PubMed]
  22. Samimi, F.; Baazm, M.; Eftekhar, E.; Rajabi, S.; Goodarzi, M.T.; Jalali Mashayekhi, F. Possible antioxidant mechanism of coenzyme Q10 in diabetes: Impact on Sirt1/Nrf2 signaling pathways. Res. Pharm. Sci. 2019, 14, 524–533. [Google Scholar] [CrossRef]
  23. Yousef, A.O.; AFahad, A.; Abdel Moneim, A.E.; Metwally, D.M.; El-Khadragy, M.F.; Kassab, R.B. The neuroprotective role of Coenzyme Q10 against lead acetate-Induced neurotoxicity Is mediated by antioxidant, anti-Inflammatory and anti-apoptotic activities. Int. J. Environ. Res. Public Health 2019, 16, 2895. [Google Scholar] [CrossRef] [PubMed]
  24. Zhao, L. Protective effects of trimetazidine and coenzyme Q10 on cisplatin-induced cardiotoxicity by alleviating oxidative stress and mitochondrial dysfunction. Anatol. J. Cardiol. 2019, 22, 232–239. [Google Scholar] [CrossRef]
  25. Sun, J.; Zhu, H.; Wang, X.; Gao, Q.; Li, Z.; Huang, H. CoQ10 ameliorates mitochondrial dysfunction in diabetic nephropathy through mitophagy. J. Endocrinol. 2019, 240, 445–465. [Google Scholar] [CrossRef]
  26. Mahmoud, A.R.; Ali, F.E.M.; Abd-Elhamid, T.H.; Hassanein, E.H.M. Coenzyme Q10 protects hepatocytes from ischemia reperfusion-induced apoptosis and oxidative stress via regulation of Bax/Bcl-2/PUMA and Nrf-2/FOXO-3/Sirt-1 signaling pathways. Tissue Cell 2019, 60, 1–13. [Google Scholar] [CrossRef] [PubMed]
  27. Li, L.; Li, H.; Zhou, Q.; Lu, Y.; Chen, P.; Wang, X.; Zhao, H. Implication of nuclear factor-erythroid 2-like 2/heme oxygenase 1 pathway in the protective effects of coenzyme Q10 against preeclampsia-like in a rat model. Microcirculation 2020, 27, e12651. [Google Scholar] [CrossRef]
  28. Al-Megrin, W.A.; Soliman, D.; Kassab, R.B.; Metwally, D.M.; Ahmed EAbdel Moneim El-Khadragy, M.F. Coenzyme Q10 activates the antioxidant machinery and inhibits the inflammatory and apoptotic cascades against lead acetate-induced renal injury in rats. Front. Physiol. 2020, 11, 64. [Google Scholar] [CrossRef]
  29. Hussein, R.M.; Sawy, D.M.; Kandeil, M.A.; Farghaly, H.S. Chlorogenic acid, quercetin, coenzyme Q10 and silymarin modulate Keap1-Nrf2/heme oxygenase-1 signaling in thioacetamide-induced acute liver toxicity. Life Sci. 2021, 277, 119460. [Google Scholar] [CrossRef]
  30. Du, Q.; Meng, W.; Athari, S.S.; Wang, R. The effect of Co-Q10 on allergic rhinitis and allergic asthma. Allergy Asthma Clin. Immunol. 2021, 17, 32. [Google Scholar] [CrossRef]
  31. Huang, J.; Zhang, H.; You, L.; Zhang, J.; Jiang, Z. Coenzyme Q10 inhibits intracranial aneurysm formation and progression in a mouse model. Pediatr. Res. 2022, 91, 839–845. [Google Scholar] [CrossRef] [PubMed]
  32. Elshamy, A.M.; Salem, O.M.; Safa, M.A.E.; Barhoma, R.A.E.; Eltabaa, E.F.; Shalaby, A.M.; Alabiad, M.A.; Arakeeb, H.M.; Mohamed, H.A. Possible protective effects of CO Q10 against vincristine-induced peripheral neuropathy: Targeting oxidative stress, inflammation, and sarmoptosis. J. Biochem. Mol. Toxicol. 2022, 36, e22976. [Google Scholar] [CrossRef] [PubMed]
  33. Tripathi, S.; Fhatima, S.; Parmar, D.; Singh, D.; Mishra, R.; Singh, G. Therapeutic effects of CoenzymeQ10, Biochanin A and Phloretin against arsenic and chromium induced oxidative stress in mouse (Mus musculus) brain. 3Biotech 2022, 121, 116. [Google Scholar] [CrossRef] [PubMed]
  34. Kaur, S.; Ahuja, P.; Kapil, L.; Sharma, D.; Singh, C.; Singh, A. Coenzyme Q10 ameliorates chemotherapy-induced cognitive impairment in mice: A preclinical study. Mol. Biol. Rep. 2024, 51, 930. [Google Scholar] [CrossRef]
  35. Deppe, L.; Mueller-Buehl, A.M.; Tsai, T.; Erb, C.; Dick, H.B.; Joachim, S.C. Protection against oxidative stress by Coenzyme Q10 in a porcine retinal degeneration model. J. Pers. Med. 2024, 14, 437. [Google Scholar] [CrossRef]
  36. Arafa, E.A.; Hassanein, E.H.M.; Ibrahim, N.A.; Buabeid, M.A.; Mohamed, W.R. Involvement of Nrf2-PPAR-γ signaling in Coenzyme Q10 protecting effect against methotrexate-induced testicular oxidative damage. Int. Immunopharmacol. 2024, 129, 111566. [Google Scholar] [CrossRef]
  37. Tripathi, S.; Parmar, D.; Singh, D.; Singh, G. Attenuation of chromium (VI) and arsenic (III)-induced oxidative stress and hepatic apoptosis by phloretin, biochanin-A, and coenzyme Q10 via activation of SIRT1/Nrf2/HO-1/NQO1 signaling. J. Biochem. Mol. Toxicol. 2024, 38, e23817. [Google Scholar] [CrossRef]
  38. Guo, Q.; Jin, Y.; Chen, X.; Ye, X.; Shen, X.; Lin, M.; Zeng, C.; Zhou, T.; Zhang, J. NF-κB in biology and targeted therapy: New insights and translational implications. Signal Transduct. Target. Ther. 2024, 9, 53. [Google Scholar] [CrossRef]
  39. Li, G.; Zou, L.Y.; Cao, C.M.; Yang, E.S. Coenzyme Q10 protects SHSY5Y neuronal cells from beta amyloid toxicity and oxygen-glucose deprivation by inhibiting the opening of the mitochondrial permeability transition pore. Biofactors 2005, 25, 97–107. [Google Scholar] [CrossRef]
  40. Brea-Calvo, G.; Siendones, E.; Sánchez-Alcázar, J.A.; de Cabo, R.; Navas, P. Cell survival from chemotherapy depends on NF-kappaB transcriptional up-regulation of coenzyme Q biosynthesis. PLoS ONE 2009, 4, e5301. [Google Scholar] [CrossRef]
  41. Kooncumchoo, P.; Sharma, S.; Porter, J.; Govitrapong, P.; Ebadi, M. Coenzyme Q(10) provides neuroprotection in iron-induced apoptosis in dopaminergic neurons. J. Mol. Neurosci. 2006, 28, 125–141. [Google Scholar] [CrossRef] [PubMed]
  42. Fouad, A.A.; Jresat, I. Hepatoprotective effect of coenzyme Q10 in rats with acetaminophen toxicity. Environ. Toxicol. Pharmacol. 2012, 33, 158–167. [Google Scholar] [CrossRef]
  43. Zhang, Y.P.; Song, C.Y.; Yuan, Y.; Eber, A.; Rodriguez, Y.; Levitt, R.C.; Takacs, P.; Yang, Z.; Goldberg, R.; Candiotti, K.A. Diabetic neuropathic pain development in type 2 diabetic mouse model and the prophylactic and therapeutic effects of coenzyme Q10. Neurobiol. Dis. 2013, 58, 169–178. [Google Scholar] [CrossRef] [PubMed]
  44. Fouad, A.A.; Al-Mulhim, A.S.; Jresat, I. Therapeutic effect of coenzyme Q10 against experimentally-induced hepatocellular carcinoma in rats. Environ. Toxicol. Pharmacol. 2013, 35, 100–108. [Google Scholar] [CrossRef]
  45. Yoneda, T.; Tomofuji, T.; Kawabata, Y.; Ekuni, D.; Azuma, T.; Kataoka, K.; Kunitomo, M.; Morita, M. Application of coenzyme Q10 for accelerating soft tissue wound healing after tooth extraction in rats. Nutrients 2014, 6, 5756–5769. [Google Scholar] [CrossRef]
  46. Li, W.; Wu, X.; Xu, X.; Wang, W.; Song, S.; Liang, K.; Yang, M.; Guo, L.; Zhao, Y.; Li, R. Coenzyme Q10 suppresses TNF-α-Induced inflammatory reaction In vitro and attenuates severity of dermatitis in mice. Inflammation 2016, 39, 281–289. [Google Scholar] [CrossRef]
  47. Li, L.; Xu, D.; Lin, J.; Zhang, D.; Wang, G.; Sui, L.; Ding, H.; Du, J. Coenzyme Q10 attenuated β-amyloid25–35-induced inflammatory responses in PC12 cells through regulation of the NF-κB signaling pathway. Brain Res. Bull. 2017, 131, 192–198. [Google Scholar] [CrossRef] [PubMed]
  48. Boroujeni, M.B.; Khayat, Z.K.; Anbari, K.; Niapour, A.; Gholami, M.; Gharravi, A.M. Coenzyme Q10 protects skeletal muscle from ischemia-reperfusion through the NF-kappa B pathway. Perfusion 2017, 32, 372–377. [Google Scholar] [CrossRef]
  49. Salehpour, F.; Farajdokht, F.; Cassano, P.; Sadigh-Eteghad, S.; Erfani, M.; Hamblin, M.R.; Salimi, M.M.; Karimi, P.; Rasta, S.H.; Mahmoudi, J. Near-infrared photobiomodulation combined with coenzyme Q10 for depression in a mouse model of restraint stress: Reduction in oxidative stress, neuroinflammation, and apoptosis. Brain Res. Bull. 2019, 144, 213–222. [Google Scholar] [CrossRef]
  50. Nyariki, J.N.; Ochola, L.A.; Jillani, N.E.; Nyamweya, N.O.; Amwayi, P.E.; Yole, D.S.; Azonvide, L.; Isaac, A.O. Oral administration of Coenzyme Q10 protects mice against oxidative stress and neuro-inflammation during experimental cerebral malaria. Parasitol. Int. 2019, 71, 106–120. [Google Scholar] [CrossRef]
  51. Quagliariello, V.; Vecchione, R.; De Capua, A.; Lagreca, E.; Iaffaioli, R.V.; Botti, G.; Netti, P.A.; Maurea, N. Nano-encapsulation of Coenzyme Q10 in secondary and tertiary nano-emulsions for enhanced cardioprotection and hepatoprotection in human cardiomyocytes and hepatocytes during exposure to anthracyclines and Ttastuzumab. Int. J. Nanomed. 2020, 15, 4859–4876. [Google Scholar] [CrossRef]
  52. Ali, F.E.M.; Ahmed, S.F.; Eltrawy, A.H.; Yousef, R.S.; Ali, H.S.; Mahmoud, A.R.; Abd-Elhamid, T.H. Pretreatment with Coenzyme Q10 combined with aescin protects against sepsis-Induced acute lung injury. Cells Tissues Organs 2021, 210, 195–217. [Google Scholar] [CrossRef]
  53. Mohamed, H.A.; Said, R.S. Coenzyme Q10 attenuates inflammation and fibrosis implicated in radiation enteropathy through suppression of NF-κB/TGF-β/MMP-9 pathways. Int. Immunopharmacol. 2021, 92, 107347. [Google Scholar] [CrossRef]
  54. Alhusaini, A.; Sarawi, W.; Mattar, D.; Abo-Hamad, A.; Almogren, R.; Alhumaidan, S.; Alsultan, E.; Alsaif, S.; Hasan, I.; Hassanein, E.; et al. Acetyl-L-carnitine and/or liposomal co-enzyme Q10 prevent propionic acid-induced neurotoxicity by modulating oxidative tissue injury, inflammation, and ALDH1A1-RA-RARα signaling in rats. Biomed. Pharmacother. 2022, 153, 113360, Corrigendum in Biomed. Pharmacother. 2023, 162, 114645. [Google Scholar] [CrossRef] [PubMed]
  55. Abd-Elhakim, Y.M.; Hashem, M.M.M.; Abo-El-Sooud, K.; Mousa, M.R.; Soliman, A.M.; Mouneir, S.M.; Ismail, S.H.; Hassan, B.A.; El-Nour, H.H.M. Interactive effects of cadmium and titanium dioxide nanoparticles on hepatic tissue in rats: Ameliorative role of coenzyme 10 via modulation of the NF-κB and TNFα pathway. Food Chem. Toxicol. 2023, 182, 114191. [Google Scholar] [CrossRef]
  56. Fakharaldeen, Z.A.; Al-Mudhafar, A.; Gany, S.N.; Radhi, A.N.; Hadi, N.R. Neuroprotective effects of Coenzyme Q10 in ischemia-reperfusion injury via inflammation and oxidative stress reduction in adult male rats. J. Med. Life. 2023, 16, 1534–1539. [Google Scholar] [CrossRef]
  57. Frontiñán-Rubio, J.; Llanos-González, E.; García-Carpintero, S.; Peinado, J.R.; Ballesteros-Yáñez, I.; Rayo, M.V.; de la Fuente, J.; Pérez-García, V.M.; Perez-Romasanta, L.A.; Malumbres, M.; et al. CoQ10 reduces glioblastoma growth and infiltration through proteome remodeling and inhibition of angiogenesis and inflammation. Cell. Oncol. 2023, 46, 65–77, Correction in Cell. Oncol. 2023, 46, 1543. [Google Scholar] [CrossRef] [PubMed]
  58. Mansour, D.F.; Hashad, I.M.; Rady, M.; Abd-El Razik, A.N.; Saleh, D.O. Diosmin and Coenzyme q10: Synergistic histopathological and functional protection against doxorubicin-induced hepatorenal injury in rats. Toxicol. Rep. 2024, 13, 101848. [Google Scholar] [CrossRef]
  59. Antar, S.A.; Abdo, W.; Helal, A.I.; Abduh, M.S.; Hakami, Z.H.; Germoush, M.O.; Alsulimani, A.; Al-Noshokaty, T.M.; El-Dessouki, A.M.; ElMahdy, M.K.; et al. Coenzyme Q10 mitigates cadmium cardiotoxicity by downregulating NF-κB/NLRP3 inflammasome axis and attenuating oxidative stress in mice. Life Sci. 2024, 348, 122688. [Google Scholar] [CrossRef]
  60. Yang, X.; Zhao, Y.; Yu, S.; Chi, L.; Cai, Y. Coenzyme Q10 alleviates neurological deficits in a mouse model of intracerebral hemorrhage by reducing inflammation and apoptosis. Exp. Biol. Med. 2025, 250, 10321. [Google Scholar] [CrossRef] [PubMed]
  61. Huang, J.; Chen, L.; Wu, J.; Ai, D.; Zhang, J.Q.; Chen, T.G.; Wang, L. Targeting the PI3K/AKT/mTOR signaling pathway in the treatment of human diseases: Current status, trends, and solutions. J. Med. Chem. 2022, 65, 16033–16061. [Google Scholar] [CrossRef]
  62. Xue, R.; Yang, J.; Wu, J.; Meng, Q.; Hao, J. Coenzyme Q10 inhibits the activation of pancreatic stellate cells through PI3K/AKT/mTOR signaling pathway. Oncotarget 2017, 8, 92300–92311. [Google Scholar] [CrossRef]
  63. Kurashiki, T.; Horikoshi, Y.; Kamizaki, K.; Sunaguchi, T.; Hara, K.; Morimoto, M.; Kitagawa, Y.; Nakaso, K.; Otsuki, A.; Matsura, T. Molecular mechanisms underlying the promotion of wound repair by coenzyme Q10: PI3K/Akt signal activation via alterations to cell membrane domains. J. Clin. Biochem. Nutr. 2022, 70, 222–230. [Google Scholar] [CrossRef]
  64. Choi, H.; Park, H.H.; Koh, S.H.; Choi, N.Y.; Yu, H.J.; Park, J.; Lee, Y.J.; Lee, K.Y. Coenzyme Q10 protects against amyloid beta-induced neuronal cell death by inhibiting oxidative stress and activating the P13K pathway. Neurotoxicology 2012, 33, 85–90. [Google Scholar] [CrossRef] [PubMed]
  65. Xu, J.; Huang, B.; Tang, S.; Sun, J.; Bao, E. Co-enzyme Q10 protects primary chicken myocardial cells from heat stress by upregulating autophagy and suppressing the PI3K/AKT/mTOR pathway. Cell Stress Chaperones 2019, 24, 1067–1078. [Google Scholar] [CrossRef]
  66. Zheng, D.; Cui, C.; Shao, C.; Wang, Y.; Ye, C.; Lv, G. Coenzyme Q10 inhibits RANKL-induced osteoclastogenesis by regulation of mitochondrial apoptosis and oxidative stress in RAW264.7 cells. J. Biochem. Mol. Toxicol. 2021, 35, e22778. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, D.; Yan, B.; Yu, S.; Zhang, C.; Wang, B.; Wang, Y.; Wang, J.; Yuan, Z.; Zhang, L.; Pan, J. Coenzyme Q10 inhibits the aging of mesenchymal stem cells induced by D-galactose through Akt/mTOR signaling. Oxidative Med. Cell Longev. 2015, 2015, 867293. [Google Scholar] [CrossRef] [PubMed]
  68. Braicu, C.; Buse, M.; Busuioc, C.; Drula, R.; Gulei, D.; Raduly, L.; Rusu, A.; Irimie, A.; Atanasov, A.G.; Slaby, O.; et al. A comprehensive review on MAPK: A promising therapeutic target in cancer. Cancers 2019, 11, 1618. [Google Scholar] [CrossRef]
  69. Li, X.; Guo, Y.; Huang, S.; He, M.; Liu, Q.; Chen, W.; Liu, M.; Xu, D.; He, P. Coenzyme Q10 prevents the interleukin-1 beta induced inflammatory response via inhibition of MAPK signaling pathways in rat articular chondrocytes. Drug Dev. Res. 2017, 78, 403–410. [Google Scholar] [CrossRef]
  70. Zheng, D.; Cui, C.; Ye, C.; Shao, C.; Zha, X.; Xu, Y.; Liu, X.; Wang, C. Coenzyme Q10 prevents RANKL-induced osteoclastogenesis by promoting autophagy via inactivation of the PI3K/AKT/mTOR and MAPK pathways. Braz. J. Med. Biol. Res. 2024, 57, e13474. [Google Scholar] [CrossRef]
  71. Allam, E.A.; Ibrahim, H.F.; Abdulmalek, S.A.; Abdelmeniem, I.M.; Basta, M. Coenzyme Q10 alleviates testicular endocrine and spermatogenic dysfunction induced by high-fat diet in male Wistar rats: Role of adipokines, oxidative stress and MAPK/ERK/JNK pathway. Andrologia 2022, 54, e14544. [Google Scholar] [CrossRef]
  72. Nawar, N.F.; Beltagy, D.M.; Tousson, E.; El-Keey, M.M.; Mohamed, T.M. Coenzyme Q10 alleviates AlCl3 and D-galactose induced Alzheimer via modulating oxidative burden and TLR-4/MAPK pathways and regulation microRNA in rat brain. Toxicol. Res. Camb 2025, 14, tfaf031. [Google Scholar] [CrossRef] [PubMed]
  73. Xu, J.; Tang, S.; Yin, B.; Sun, J.; Bao, E. Co-enzyme Q10 upregulates Hsp70 and protects chicken primary myocardial cells under in vitro heat stress via PKC/MAPK. Mol. Cell. Biochem. 2018, 449, 195–206. [Google Scholar] [CrossRef]
  74. Hu, Q.; Bian, Q.; Rong, D.; Wang, L.; Song, J.; Huang, H.S.; Zeng, J.; Mei, J.; Wang, P.Y. JAK/STAT pathway: Extracellular signals, diseases, immunity, and therapeutic regimens. Front. Bioeng. Biotechnol. 2023, 11, 1110765. [Google Scholar] [CrossRef] [PubMed]
  75. Schmelzer, C.; Lindner, I.; Vock, C.; Fujii, K.; Döring, F. Functional connections and pathways of coenzyme Q10-inducible genes: An in-silico study. IUBMB Life 2007, 59, 628–633. [Google Scholar] [CrossRef]
  76. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  77. Hussein, Z.; Michel, H.E.; El-Naga, R.N.; El-Demerdash, E.; Mantawy, E.M. Coenzyme Q10 ameliorates cyclophosphamide-induced chemobrain by repressing neuronal apoptosis and preserving hippocampal neurogenesis: Mechanistic roles of Wnt/ β-catenin signaling pathway. Neurotoxicology 2024, 105, 21–33. [Google Scholar] [CrossRef]
  78. Jiang, Y.J.; Jin, J.; Nan, Q.Y.; Ding, J.; Cui, S.; Xuan, M.Y.; Piao, M.H.; Piao, S.G.; Zheng, H.L.; Jin, J.Z.; et al. Coenzyme Q10 attenuates renal fibrosis by inhibiting RIP1-RIP3-MLKL-mediated necroinflammation via Wnt3α/β-catenin/GSK-3β signaling in unilateral ureteral obstruction. Int. Immunopharmacol. 2022, 108, 108868. [Google Scholar] [CrossRef] [PubMed]
  79. Ozhan, G.; Weidinger, G. Wnt/β-catenin signaling in heart regeneration. Cell Regen. 2015, 4, 3. [Google Scholar] [CrossRef]
  80. Mortensen, S.A.; Rosenfeldt, F.; Kumar, A.; Dolliner, P.; Filipiak, K.J.; Pella, D.; Alehagen, U.; Steurer, G.; Littarru, G.P.; Q-SYMBIO Study Investigators. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: Results from Q-SYMBIO: A randomized double-blind trial. JACC Heart Fail. 2014, 2, 641–649. [Google Scholar] [CrossRef]
  81. Herzig, S.; Shaw, R.J. AMPK: Guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 2018, 19, 121–135. [Google Scholar] [CrossRef] [PubMed]
  82. Xie, T.; Wang, C.; Jin, Y.; Meng, Q.; Liu, Q.; Wu, J.; Sun, H. CoenzymeQ10-induced activation of AMPK-YAP-OPA1 pathway alleviates atherosclerosis by improving mitochondrial function, inhibiting oxidative stress and promoting energy metabolism. Front. Pharmacol. 2020, 11, 1034. [Google Scholar] [CrossRef]
  83. Robbins, D.J.; Fei, D.L.; Riobo, N.A. The Hedgehog signal transduction network. Sci. Signal. 2012, 5, re6. [Google Scholar] [CrossRef]
  84. Patel, S.; Armbruster, H.; Pardo, G.; Archambeau, B.; Kim, N.H.; Jeter, J.; Wu, R.; Kendra, K.; Contreras, C.M.; Spaccarelli, N.; et al. Hedgehog pathway inhibitors for locally advanced and metastatic basal cell carcinoma: A real-world single-center retrospective review. PLoS ONE 2024, 19, e0297531. [Google Scholar] [CrossRef]
  85. Govorova, I.A.; Nikitochkina, S.Y.; Vorotelyak, E.A. Influence of intersignaling crosstalk on the intracellular localization of YAP/TAZ in lung cells. Cell Commun. Signal. 2024, 22, 289. [Google Scholar] [CrossRef]
  86. Zeke, A.; Lukács, M.; Lim, W.A.; Reményi, A. Scaffolds: Interaction platforms for cellular signalling circuits. Trends Cell Biol. 2009, 19, 364–374. [Google Scholar] [CrossRef]
  87. Aksamitiene, E.; Kiyatkin, A.; Kholodenko, B.N. Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt pathways: A fine balance. Biochem. Soc. Trans. 2012, 40, 139–146. [Google Scholar] [CrossRef]
  88. Mantle, D.; Rowbottom, H.; Jones, J.; Potts, I.M.; Turton, N.; Dewsbury, M.; Lopez-Lluch, G.; Hargreaves, I.P. Energy metabolism as a therapeutic target in cancer: The role of coenzyme Q10. Oxygen 2024, 4, 122–138. [Google Scholar] [CrossRef]
  89. Lee, S.K.; Lee, J.O.; Kim, J.H.; Kim, N.; You, G.Y.; Moon, J.W.; Sha, J.; Kim, S.J.; Lee, Y.W.; Kang, H.J.; et al. Coenzyme Q10 increases the fatty acid oxidation through AMPK-mediated PPARα induction in 3T3-L1 preadipocytes. Cell Signal. 2012, 24, 2329–2336. [Google Scholar] [CrossRef] [PubMed]
  90. Johnson, D.A.; Johnson, J.A. Nrf2—A therapeutic target for the treatment of neurodegenerative diseases. Free Radic. Biol. Med. 2015, 88 Pt B, 253–267. [Google Scholar] [CrossRef]
  91. Sun, S.C.; Chang, J.H.; Jin, J. Regulation of nuclear factor-κB in autoimmunity. Trends Immunol. 2013, 34, 282–289. [Google Scholar] [CrossRef] [PubMed]
  92. Rascio, F.; Spadaccino, F.; Rocchetti, M.T.; Castellano, G.; Stallone, G.; Netti, G.S.; Ranieri, E. The pathogenic role of PI3K/AKT pathway in cancer onset and drug resistance: An updated review. Cancers 2021, 13, 3949. [Google Scholar] [CrossRef]
  93. Bahar, M.E.; Kim, H.J.; Kim, D.R. Targeting the RAS/RAF/MAPK pathway for cancer therapy: From mechanism to clinical studies. Signal Transduct. Target. Ther. 2023, 8, 455. [Google Scholar] [CrossRef] [PubMed]
  94. Parveen, S.; Fatma, M.; Mir, S.S.; Dermime, S.; Uddin, S. JAK-STAT signaling in autoimmunity and cancer. Immunotargets Ther. 2025, 14, 523–554. [Google Scholar] [CrossRef]
  95. Sidrat, T.; Rehman, Z.U.; Joo, M.D.; Lee, K.L.; Kong, I.K. Wnt/β-catenin pathway-mediated PPARδ expression during embryonic development differentiation and disease. Int. J. Mol. Sci. 2021, 22, 1854. [Google Scholar] [CrossRef]
  96. Wu, S.; Zou, M.H. AMPK, Mitochondrial function, and cardiovascular disease. Int. J. Mol. Sci. 2020, 21, 4987. [Google Scholar] [CrossRef]
  97. Abramyan, J. Hedgehog signaling and embryonic craniofacial disorders. J. Dev. Biol. 2019, 7, 9. [Google Scholar] [CrossRef]
  98. Ramsey, C.P.; Glass, C.A.; Montgomery, M.B.; Lindl, K.A.; Ritson, G.P.; Chia, L.A.; Hamilton, R.L.; Chu, C.T.; Jordan-Sciutto, K.L. Expression of Nrf2 in neurodegenerative diseases. J. Neuropathol. Exp. Neurol. 2007, 66, 75–85. [Google Scholar] [CrossRef]
  99. Yang, X.X.; Yang, R.; Zhang, F. Role of Nrf2 in Parkinson’s Disease: Toward new perspectives. Front. Pharmacol. 2022, 13, 919233. [Google Scholar] [CrossRef] [PubMed]
  100. Sarlette, A.; Krampfl, K.; Grothe, C.; Neuhoff Nv Dengler, R.; Petri, S. Nuclear erythroid 2-related factor 2-antioxidative response element signaling pathway in motor cortex and spinal cord in amyotrophic lateral sclerosis. J. Neuropathol. Exp. Neurol. 2008, 67, 1055–1062. [Google Scholar] [CrossRef]
  101. Quinti, L.; Naidu, S.D.; Träger, U.; Chen, X.; Kegel-Gleason, K.; Llères, D.; Connolly, C.; Chopra, V.; Low, C.; Moniot, S.; et al. KEAP1-modifying small molecule reveals muted NRF2 signaling responses in neural stem cells from Huntington’s disease patients. Proc. Natl. Acad. Sci. USA 2017, 114, E4676–E4685. [Google Scholar] [CrossRef]
  102. Petrillo, S.; D’Amico, J.; La Rosa, P.; Bertini, E.S.; Piemonte, F. Targeting NRF2 for the treatment of Friedreich’s Ataxia: A comparison among drugs. Int. J. Mol. Sci. 2019, 20, 5211. [Google Scholar] [CrossRef]
  103. Khan, S.U.; Khan, S.U.; Suleman, M.; Khan, M.U.; Khan, M.S.; Arbi, F.M.; Hussain, T.; Mohammed Alsuhaibani, A.; SRefat, M. Natural allies for heart health: Nrf2 activation and cardiovascular disease management. Curr. Probl. Cardiol. 2024, 49 Pt B, 102084. [Google Scholar] [CrossRef]
  104. Liu, P.; Anandhan, A.; Chen, J.; Shakya, A.; Dodson, M.; Ooi, A.; Chapman, E.; White, E.; Garcia, J.G.; Zhang, D.D. Decreased autophagosome biogenesis, reduced NRF2, and enhanced ferroptotic cell death are underlying molecular mechanisms of non-alcoholic fatty liver disease. Redox Biol. 2023, 59, 102570. [Google Scholar] [CrossRef] [PubMed]
  105. Juul-Nielsen, C.; Shen, J.; Stenvinkel, P.; Scholze, A. Systematic review of the nuclear factor erythroid 2-related factor 2 (NRF2) system in human chronic kidney disease: Alterations, interventions and relation to morbidity. Nephrol. Dial. Transplant. 2022, 37, 904–916. [Google Scholar] [CrossRef] [PubMed]
  106. Ban, W.H.; Rhee, C.K. Role of Nuclear Factor Erythroid 2–Related Factor 2 in chronic obstructive pulmonary disease. Tuberc. Respir. Dis. Seoul 2022, 85, 221–226. [Google Scholar] [CrossRef] [PubMed]
  107. Wen, Z.; Liu, W.; Li, X.; Chen, W.; Liu, Z.; Wen, J.; Liu, Z. A protective role of the NRF2-Keap1 pathway in maintaining Intestinal barrier function. Oxidative Med. Cell. Longev. 2019, 2019, 1759149. [Google Scholar] [CrossRef]
  108. Zhang, X.; Li, H.; Chen, L.; Wu, Y.; Li, Y. NRF2 in age-related musculoskeletal diseases: Role and treatment prospects. Genes Dis. 2023, 11, 101180. [Google Scholar] [CrossRef]
  109. Paplomata, E.; Zelnak, A.; Santa-Maria, C.A.; Liu, Y.; Gogineni, K.; Li, X.; Moreno, C.S.; Chen, Z.; Kaklamani, V.; O’Regan, R.M. Use of Everolimus and Trastuzumab in addition to endocrine therapy in hormone-refractory metastatic breast cancer. Clin. Breast Cancer 2019, 19, 188–196. [Google Scholar] [CrossRef]
  110. Lynch, D.R.; Chin, M.P.; Delatycki, M.B.; Subramony, S.H.; Corti, M.; Hoyle, J.C.; Boesch, S.; Nachbauer, W.; Mariotti, C.; Mathews, K.D.; et al. Safety and efficacy of Omaveloxolone in Friedreich Ataxia (MOXIe Study). Ann. Neurol. 2021, 89, 212–225, Correction in Ann. Neurol. 2023, 94, 1190. [Google Scholar] [CrossRef]
  111. Okuda, D.T.; Kantarci, O.; Lebrun-Frénay, C.; Sormani, M.P.; Azevedo, C.J.; Bovis, F.; Hua, L.H.; Amezcua, L.; Mowry, E.M.; Hotermans, C.; et al. Dimethyl fumarate delays multiple sclerosis in radiologically isolated syndrome. Ann. Neurol. 2023, 93, 604–614. [Google Scholar] [CrossRef]
  112. Cengiz Seval, G.; Beksac, M. The safety of bortezomib for the treatment of multiple myeloma. Expert Opin. Drug Saf. 2018, 17, 953–962. [Google Scholar] [CrossRef]
  113. Kavanaugh, S.M.; White, L.A.; Kolesar, J.M. Vorinostat: A novel therapy for the treatment of cutaneous T-cell lymphoma. Am. J. Health Syst. Pharm. 2010, 67, 793–797. [Google Scholar] [CrossRef]
  114. Copur, M.S. Alpelisib to treat breast cancer. Drugs Today 2020, 56, 357–363. [Google Scholar] [CrossRef] [PubMed]
  115. Santin, A.D.; Filiaci, V.; Bellone, S.; Ratner, E.S.; Mathews, C.A.; Cantuaria, G.; Gunderson, C.C.; Rutledge, T.; Buttin, B.M.; Lankes, H.A.; et al. Phase II evaluation of copanlisib, a selective inhibitor of Pi3kca, in patients with persistent or recurrent endometrial carcinoma harboring PIK3CA hotspot mutations: An NRG Oncology study (NRG-GY008). Gynecol. Oncol. Rep. 2020, 31, 100532, Corrigendum in Gynecol. Oncol. Rep. 2020, 33, 100590. [Google Scholar] [CrossRef] [PubMed]
  116. Turner, N.C.; Oliveira, M.; Howell, S.J.; Dalenc, F.; Cortes, J.; Moreno, H.L.G.; Hu, X.; Jhaveri, K.; Krivorotko, P.; Loibl, S.; et al. Capivasertib in hormone receptor-positive advanced breast cancer. N. Engl. J. Med. 2023, 388, 2058–2070. [Google Scholar] [CrossRef]
  117. Sutaria, D.S.; Rasuo, G.; Harris, A.; Johnson, R.; Miles, D.; Gallo, J.D.; Sane, R. Drug-drug interaction study to evaluate the pharmacokinetics, safety, and tolerability of Ipatasertib in combination with Darolutamide in patients with advanced prostate cancer. Pharmaceutics 2022, 14, 2101. [Google Scholar] [CrossRef]
  118. Amato, R.; Stepankiw, M. Evaluation of everolimus in renal cell cancer. Expert Opin. Pharmacother. 2013, 14, 1229–1240. [Google Scholar] [CrossRef]
  119. Goudarzi, Z.; Mostafavi, M.; Salesi, M.; Jafari, M.; Mirian, I.; Hashemi Meshkini, A.; Keshavarz, K.; Ghasemi, Y. Everolimus and temsirolimus are not the same second-line in metastatic renal cell carcinoma: A systematic review and meta-analysis. Cost. Eff. Resour. Alloc. 2023, 21, 10. [Google Scholar] [CrossRef]
  120. Aires-Lopes, B.; Justo, G.Z.; Cordeiro, H.G.; Durán, N.; Azevedo-Martins, J.M.; Ferreira Halder, C.V. Violacein improves vemurafenib response in melanoma spheroids. Nat. Prod. Res. 2024, 38, 3417–3420. [Google Scholar] [CrossRef] [PubMed]
  121. Long, G.V.; Hauschild, A.; Santinami, M.; Kirkwood, J.M.; Atkinson, V.; Mandala, M.; Merelli, B.; Sileni, V.C.; Nyakas, M.; Haydon, A.; et al. Final results for adjuvant Dabrafenib plus Trametinib in Stage III melanoma. N. Engl. J. Med. 2024, 391, 1709–1720. [Google Scholar] [CrossRef] [PubMed]
  122. Thota, R.; Johnson, D.B.; Sosman, J.A. Trametinib in the treatment of melanoma. Expert Opin. Biol. Ther. 2015, 15, 735–747. [Google Scholar] [CrossRef] [PubMed]
  123. Kim, H.; Yoon, H.M.; Kim, E.K.; Ra, Y.S.; Kim, H.W.; Yum, M.S.; Kim, M.J.; Baek, J.S.; Sung, Y.S.; Lee, S.M.; et al. Safety and efficacy of selumetinib in pediatric and adult patients with neurofibromatosis type 1 and plexiform neurofibroma. Neuro Oncol. 2024, 26, 2352–2363. [Google Scholar] [CrossRef]
  124. Urits, I.; Israel, J.; Hakobyan, H.; Yusin, G.; Lassiter, G.; Fackler, N.; Berger, A.A.; Kassem, H.; Kaye, A.; Viswanath, O. Baricitinib for the treatment of rheumatoid arthritis. Reumatologia 2020, 58, 407–415. [Google Scholar] [CrossRef] [PubMed]
  125. Simpson, E.L.; Papp, K.A.; Blauvelt, A.; Chu, C.Y.; Hong, H.C.; Katoh, N.; Calimlim, B.M.; Thyssen, J.P.; Chiou, A.S.; Bissonnette, R.; et al. Efficacy and safety of Upadacitinib in patients with moderate to severe atopic dermatitis: Analysis of follow-up data from the Measure Up 1 and Measure Up 2 randomized clinical trials. JAMA Dermatol. 2022, 158, 404–413. [Google Scholar] [CrossRef]
  126. Oh, D.Y.; Lee, S.H.; Han, S.W.; Kim, M.J.; Kim, T.M.; Kim, T.Y.; Heo, D.S.; Yuasa, M.; Yanagihara, Y.; Bang, Y.J. Phase I study of OPB-31121, an oral STAT3 inhibitor, in patients with advanced solid tumors. Cancer Res. Treat. 2015, 47, 607–615. [Google Scholar] [CrossRef]
  127. Aguilar-Recarte, D.; Barroso, E.; Zhang, M.; Rada, P.; Pizarro-Delgado, J.; Peña, L.; Palomer, X.; Valverde, Á.M.; Wahli, W.; Vázquez-Carrera, M. A positive feedback loop between AMPK and GDF15 promotes metformin antidiabetic effects. Pharmacol. Res. 2023, 187, 106578. [Google Scholar] [CrossRef]
  128. Zhang, J.; Nannapaneni, S.; Wang, D.; Liu, F.; Wang, X.; Jin, R.; Liu, X.; Rahman, M.A.; Peng, X.; Qian, G.; et al. Phenformin enhances the therapeutic effect of selumetinib in KRAS-mutant non-small cell lung cancer irrespective of LKB1 status. Oncotarget 2017, 8, 59008–59022. [Google Scholar] [CrossRef]
  129. Sekulic, A.; Migden, M.R.; Oro, A.E.; Dirix, L.; Lewis, K.D.; Hainsworth, J.D.; Solomon, J.A.; Yoo, S.; Arron, S.T.; Friedlander, P.A.; et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N. Engl. J. Med. 2012, 366, 2171–2179. [Google Scholar] [CrossRef]
  130. Fersing, C.; Mathias, F. Update on glasdegib in acute myeloid leukemia—Broadening horizons of Hedgehog pathway inhibitors. Acta Pharm. 2021, 72, 9–34. [Google Scholar] [CrossRef]
  131. Wang, Q.; Chaerkady, R.; Wu, J.; Hwang, H.J.; Papadopoulos, N.; Kopelovich, L.; Maitra, A.; Matthaei, H.; Eshleman, J.R.; Hruban, R.H.; et al. Mutant proteins as cancer-specific biomarkers. Proc. Natl. Acad. Sci. USA 2011, 108, 2444–2449. [Google Scholar] [CrossRef]
  132. Abarca-Zabalía, J.; García, M.I.; Lozano Ros, A.; Marín-Jiménez, I.; Martínez-Ginés, M.L.; López-Cauce, B.; Martín-Barbero, M.L.; Salvador-Martín, S.; Sanjurjo-Saez, M.; García-Domínguez, J.M.; et al. Differential expression of SMAD genes and S1PR1 on circulating CD4+ T cells in multiple sclerosis and Crohn’s Disease. Int. J. Mol. Sci. 2020, 21, 676. [Google Scholar] [CrossRef]
  133. Scuto, M.; Majzúnová, M.; Torcitto, G.; Antonuzzo, S.; Rampulla, F.; Di Fatta, E.; Trovato Salinaro, A. Functional Food Nutrients, Redox Resilience Signaling and Neurosteroids for Brain Health. Int. J. Mol. Sci. 2024, 25, 12155. [Google Scholar] [CrossRef]
  134. Scuto, M.C.; Anfuso, C.D.; Lombardo, C.; Di Fatta, E.; Ferri, R.; Musso, N.; Zerbo, G.; Terrana, M.; Majzúnová, M.; Lupo, G.; et al. Neuronutrition and Nrf2 Brain Resilience Signaling: Epigenomics and Metabolomics for Personalized Medicine in Nervous System Disorders from Bench to Clinic. Int. J. Mol. Sci. 2025, 26, 9391. [Google Scholar] [CrossRef]
  135. Ferrante, K.L.; Shefner, J.; Zhang, H.; Betensky, R.; O’Brien, M.; Yu, H.; Fantasia, M.; Taft, J.; Beal, M.F.; Traynor, B.; et al. Tolerance of high-dose (3,000 mg/day) coenzyme Q10 in ALS. Neurology 2005, 65, 1834–1836. [Google Scholar] [CrossRef] [PubMed]
  136. Mantle, D.; Hargreaves, I.P. Mitochondrial dysfunction and neurodegenerative disorders: Role of nutritional supplementation. Int. J. Mol. Sci. 2022, 23, 12603. [Google Scholar] [CrossRef]
  137. Mantle, D.; Hargreaves, I.P. Disorders of human Coenzyme Q10 metabolism: An overview. Int. J. Mol. Sci. 2024, 25, 4576. [Google Scholar] [CrossRef]
  138. Mantle, D.; Heaton, R.A.; Hargreaves, I.P. Coenzyme Q10 and immune function: An overview. Antioxidants 2021, 10, 759. [Google Scholar] [CrossRef] [PubMed]
  139. Mantle, D.; Kozhevnikova, S.; Larsen, S. Coenzyme Q10 and obesity: An overview. Antioxidants 2025, 14, 871. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effect of coenzyme Q10 on cellular signalling pathways.
Figure 1. Effect of coenzyme Q10 on cellular signalling pathways.
Ijms 26 11024 g001
Table 1. Summary of studies investigating the effect of CoQ10 on the Nrf2 pathway.
Table 1. Summary of studies investigating the effect of CoQ10 on the Nrf2 pathway.
Outcome of CoQ10 SupplementationSystem StudiedStudy
Nrf2 increased, antioxidant enzymes increased, fibrosis decreased.Fibrogenesis in mouse liverChoi et al. [16]
Nrf2 increased, inflammation decreased.Exercise training in ratsPala et al. [17]
Nrf2 increased, oxidative stress reduced, inflammation reduced.Liver inflammation in miceYang et al. [18]
Nrf2 increased, antioxidant enzymes increased, cell viability increased.Neurotoxicity in PC12 cellsLi et al. [19]
Nrf2 increased, antioxidant enzymes increased, improved renal function.Nephrotoxicity in miceKabel & Elkhoely [20]
Nrf2 increased, decreased oxidative stress, inflammation, and apoptosis.Ulcerative colitis in ratsKhodir et al. [21]
Nrf2 increased, oxidative stress decreased.Diabetes in ratsSamimi et al. [22]
Nrf2 increased, oxidative stress and apoptosis decreased.Spinal cord injury in ratsLi et al. [15]
Nrf2 increased, oxidative stress, inflammation, and apoptosis decreased.Lead induced neurotoxicity in ratsYousef et al. [23]
Nrf2 increased, oxidative stress, and apoptosis decreased, improved cell viability.Cisplatin induced toxicity in rat cardiomyocytesZhao [24]
Nrf2 increased, oxidative stress decreased.Diabetic nephropathy in miceSun et al. [25]
Nrf2 increased, oxidative stress, inflammation, and apoptosis reduced.Hepatic ischaemia–reperfusion injury in ratsMahmoud et al. [26]
Nrf2 increased, oxidative stress and inflammation reduced, decreased blood pressure.Pre-eclampsia in ratsLi et al. [27]
Nrf2 increased, oxidative stress, inflammation and apoptosis reduced.Lead induced nephrotoxicity in ratsAl-Megrin et al. [28]
Nrf2 increased, oxidative stress, inflammation, and apoptosis reduced, improved liver function.Thioacetamide induced liver toxicity in ratsHussein et al. [29]
Nrf2 increased, oxidative stress, inflammation, and apoptosis reduced, allergy status improved.Allergic rhinitis/asthma in miceDu et al. [30]
Nrf2 increased, oxidative stress and apoptosis decreased, aneurysm formation reduced.Intracranial aneurysm in miceHuang et al. [31]
Nrf2 increased, oxidative stress and inflammation reduced.Vincristine induced peripheral neuropathy in ratsElsamy et al. [32]
Nrf2 increased, oxidative stress reduced.Arsenic/chromium neurotoxicity in miceTripathy et al. [33]
Nrf2 increased, oxidative stress and inflammation decreased, cognition improved.Chemotherapy induced cognitive impairment in miceKaur et al. [34]
Nrf2 increased, oxidative stress decreased, loss of retinal cells reduced.Porcine retinal explant degenerationDeppe et al. [35]
Nrf2 increased, oxidative stress decreased.Diabetes in ratsSamimi et al. [13]
Nrf2 increased, oxidative stress, inflammation, and apoptosis reduced.Testicular damage in ratsArafa et al. [36]
Nrf2 increased, oxidative stress and apoptosis decreased.Arsenic/chromium hepatotoxicity in miceTripathi et al. [37]
Table 2. Summary of studies investigating the effect of CoQ10 on the NF-κB pathway.
Table 2. Summary of studies investigating the effect of CoQ10 on the NF-κB pathway.
Outcome of CoQ10 SupplementationSystem StudiedStudy
NF-κB downregulated, oxidative stress, inflammation and apoptosis reducedMouse model of Parkinson’s diseaseKooncumchoo et al. [41]
NF-κB downregulated, oxidative stress, inflammation and apoptosis reducedAcetaminophen induced liver toxicity in ratsFouad et al. [42]
NF-κB downregulated, oxidative stress reducedNeuropathic pain in diabetic miceZhang et al. [43]
NF-κB downregulated, oxidative stress and inflammation reducedTrichloroacetic acid induced hepatocellular carcinoma in ratsFouad et al. [44]
NF-κB downregulated, inflammation reducedWound healing in ratsYoneda et al. [45]
NF-κB downregulated, inflammation reducedOxazolone induced dermatitis in miceLi et al. [46]
NF-κB downregulated, inflammation reducedAmyloid induced inflammation in PC12 cellsLi et al. [47]
NF-κB downregulated, inflammation reducedIschaemia–reperfusion injury in skeletal muscle of ratsBoroujeni et al. [48]
NF-κB downregulated, oxidative stress, inflammation and apoptosis reducedRestraint induced depression in miceSalehpour et al. [49]
NF-κB downregulated, oxidative stress and inflammation reducedCerebral malaria in miceNyariki et al. [50]
NF-κB downregulated, oxidative stress and inflammation reducedAnthracycline induced toxicity in human cardiomyocytesQuagliariello et al. [51]
NF-κB downregulated, mitochondrial function improved, inflammation reducedLipopolysaccharide induced lung injury in ratsAli et al. [52]
NF-κB downregulated, oxidative stress and inflammation reducedRadiation induced enteropathy in ratsMohamed & Said [53]
NF-κB downregulated, oxidative stress, inflammation and apoptosis reducedPropionic acid induced cerebral injury in ratsAlhusaini et al. [54]
NF-κB downregulated, oxidative stress and inflammation reducedCadmium/titanium induced liver toxicity in ratsAbd-Elkahim et al. [55]
NF-κB downregulated, oxidative stress and inflammation reducedCerebral ischaemia–reperfusion injury in ratsFakharaldeen et al. [56]
NF-κB downregulated, tumour cell invasiveness decreasedGlioblastoma in miceFrontinan-Rubio et al. [57]
NF-κB downregulated, oxidative stress and inflammation reducedDoxorubicin induced liver toxicity in ratsMansour et al. [58]
NF-κB downregulated, oxidative stress and inflammation reducedCadmium cardiotoxicity in ratsAntar et al. [59]
NF-κB downregulated, inflammation and apoptosis reducedIntracerebral haemorrhage in miceYang et al. [60]
Table 4. Medicines targeting intracellular signalling pathways.
Table 4. Medicines targeting intracellular signalling pathways.
PathwayMedicineDisorderReference
Nrf2/NQO1Nrf2 activators:
omaveloxolone
dimethyl fumarate

Friedreich’s ataxia
multiple sclerosis

Lynch et al. [110]
Okuda et al. [111]
NF-κBNF-κB inhibitors:
bortezomib
vorinostat

myeloma
cutaneous T-cell lymphoma

Cengiz-Seval et al. [112]
Kavanaugh et al. [113]
P13K/AKT/mTORP13K inhibitors:
alpelisib
copanlisib
AKT inhibitors:
capivasertib
ipatasertib
mTOR inhibitors:
everolimus
temsirolimus

breast cancer
endometrial cancer

breast, prostate cancer
prostate cancer

renal cell cancer
renal cell cancer

Copur [114]
Santin et al. [115]

Turner et al. [116]
Sutaria et al. [117]

Amato [118]
Goudarzi et al. [119]
MAPKRAF inhibitors:
vemurafenib
dabrafenib
MEK inhibitors:
trametinib
selumetinib

melanoma
melanoma

melanoma
neurofibroma

Aires-Lopez [120]
Long et al. [121]

Thota et al. [122]
Kim et al. [123]
JAK/STATJAK inhibitors:
baricitinib
upadacitinib
STAT inhibitors:
OBP-31121

rheumatoid arthritis
atopic dermatitis

various cancers

Urits et al. [124]
Simpson et al. [125]

Oh et al. [126]
AMPK/YAP/OPA1AMPK activators:
metformin
phenformin

type II diabetes
cancer

Aguilar-Recarte [127]
Zhang et al. [128]
HedgehogSMO inhibitors:
vismodegib
glasdegib

basal cell carcinoma
leukaemia

Sekulic et al. [129]
Fersing [130]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mantle, D. Coenzyme Q10 and Intracellular Signalling Pathways: Clinical Relevance. Int. J. Mol. Sci. 2025, 26, 11024. https://doi.org/10.3390/ijms262211024

AMA Style

Mantle D. Coenzyme Q10 and Intracellular Signalling Pathways: Clinical Relevance. International Journal of Molecular Sciences. 2025; 26(22):11024. https://doi.org/10.3390/ijms262211024

Chicago/Turabian Style

Mantle, David. 2025. "Coenzyme Q10 and Intracellular Signalling Pathways: Clinical Relevance" International Journal of Molecular Sciences 26, no. 22: 11024. https://doi.org/10.3390/ijms262211024

APA Style

Mantle, D. (2025). Coenzyme Q10 and Intracellular Signalling Pathways: Clinical Relevance. International Journal of Molecular Sciences, 26(22), 11024. https://doi.org/10.3390/ijms262211024

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop